Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/236458105

Effect of maternal micronutrients (folic acid and vitamin B12) and omega 3
fatty acids on indices of brain oxidative stress in the offspring

Article  in  Brain & Development · April 2013


DOI: 10.1016/j.braindev.2013.03.004 · Source: PubMed

CITATIONS READS

27 195

6 authors, including:

Suchitra Roy Amrita Ankush Khaire


Bharati Vidyapeeth Deemed University Bharati Vidyapeeth Deemed University
9 PUBLICATIONS   182 CITATIONS    41 PUBLICATIONS   337 CITATIONS   

SEE PROFILE SEE PROFILE

Karuna Randhir Anvita Kale

13 PUBLICATIONS   215 CITATIONS   
Interactive Research School for Health Affairs, Bharati Vidyapeeth Deemed Univer…
94 PUBLICATIONS   1,608 CITATIONS   
SEE PROFILE
SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Maternal micronutrients, omega 3 fatty acids and gene expression of angiogenic and inflammatory markers in pregnancy induced hypertension rats. View project

Epigenetic View project

All content following this page was uploaded by Anvita Kale on 21 August 2018.

The user has requested enhancement of the downloaded file.


Brain & Development 34 (2012) 64–71
www.elsevier.com/locate/braindev

Original article

Maternal micronutrients (folic acid and vitamin B12) and omega


3 fatty acids: Implications for neurodevelopmental
risk in the rat offspring
Suchitra Roy 1, Anvita Kale 1, Kamini Dangat, Pratiksha Sable,
Asmita Kulkarni, Sadhana Joshi ⇑
Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune 411043, India

Received 15 September 2010; received in revised form 5 January 2011; accepted 8 January 2011

Abstract

Altered maternal micronutrients (folic acid, vitamin B12) are suggested to be at the heart of intra-uterine programming of adult
diseases. We have recently described interactions of folic acid, vitamin B12 and docosahexaenoic acid in one carbon metabolism that
is considered to play a key role in regulation oxidative stress and chromatin methylation. However its impact on fetal oxidative stress
and brain fatty acid levels has been relatively unexplored. The present study examined the effect of imbalance in maternal micro-
nutrients (folic acid and vitamin B12) and maternal omega 3 fatty acid supplementation on oxidative stress parameters and brain
fatty acids and in the offspring at birth. Pregnant female rats were divided into six groups at two levels of folic acid both in the
presence and absence of vitamin B12. Both the vitamin B12 deficient groups were supplemented with omega 3 fatty acid. Oxidative
stress marker (malondialdehyde) and polyunsaturated fatty acid profiles in plasma and brain were analyzed in dam and offspring at
d20. Our results for the first time indicate that imbalance in maternal micronutrients (excess maternal folic acid supplementation on
a B12 deficient diet) increases (p < 0.01) oxidative stress in both mother and pups. This increased maternal oxidative stress resulted in
lower (p < 0.01) fetal brain DHA levels. Omega 3 fatty acid supplementation was able to restore (p < 0.05) the levels of brain DHA
in both the vitamin B12 deficient groups. Our data has implications for implications for neurodevelopmental disorders since micro-
nutrients and DHA are important modulators for neural functioning.
Ó 2011 The Japanese Society of Child Neurology. Published by Elsevier B.V. All rights reserved.

Keywords: Docosahexaenoic acid; Folic acid; Micronutrients; Oxidative stress; Vitamin B12

1. Introduction developmental origins of disease has been on the avail-


ability of energy and protein during these critical devel-
It is well established that the environment encoun- opmental periods, micronutrient deficiencies may also
tered during fetal life and infancy is strongly related play an important role in fetal growth and development
to increased risk of non-communicable diseases in [2]. In developing countries, like India micronutrient
adult life [1]. Although much of the focus on the deficiencies are common and associated with poor
pregnancy outcomes [3]. Importance of maternal folic
acid in prevention of neural tube defects is well estab-
⇑ Corresponding author. Address: Interactive Research School for
lished [4]. In this view, in India, there is a policy for
Health Sciences, Bharati Vidyapeeth University, Pune Satara Road,
supplementing all pregnant women with iron and folic
Pune 411043, India. Tel.: +91 020 24366929x31; fax: +91 020 24366929.
E-mail address: srjoshi62@gmail.com (S. Joshi). acid (60 mg and 500 lg per day) for prevention of neural
1
These authors contributed equally to this work. tube defects [5].

0387-7604/$ - see front matter Ó 2011 The Japanese Society of Child Neurology. Published by Elsevier B.V. All rights reserved.
doi:10.1016/j.braindev.2011.01.002
S. Roy et al. / Brain & Development 34 (2012) 64–71 65

Many countries like USA, Canada and Chile have of control and supervision of experimental animals,
also introduced folic acid fortification program wherein Govt of India (No. 258/CPCSEA).
all flour and uncooked cereal-grain products were forti-
fied with folic acid [6–9]. This folic acid fortification has 2.1. Animals
been shown to improve the folate status and reduce the
prevalence and incidence of neural tube defects [10–15]. Wistar albino rats (60F, 15M) of average weight 150 g
However, the long-term effect of folic acid fortification were obtained from National Toxicology Center animal
on the risk of colorectal cancer and other malignancies house. Instead of using them directly for the experimental
remain unclear [16]. Recent studies have raised concerns protocol it was thought appropriate to use their progeny.
that folic acid fortification may mask symptoms as well They were maintained at 22 °C on a controlled 12-h light
as worsen the vitamin B12 deficiency and lead to adverse and 12 h dark cycle with appropriate ventilation system.
neurologic consequences [17,18]. Most of the Indian Animals were marked with picric acid as Head (H), Back
women during pregnancy are known to have lower vita- (B), Tail (T), etc. for identification.
min B12 levels [19] and is therefore a cause of concern.
Our own studies in animals and by others have shown 2.2. Breeding
that folic acid alters levels of omega 3 fatty acids [20]. A
series of our studies in pregnancy have well established These pups were then put for breeding at 3 months of
the importance of omega 3 fatty acids especially DHA age. Males were housed individually prior to mating to
during pregnancy [21–25]. Docosahexaenoic acid acquire cage dominance. Virgin female rats were
(DHA), an omega 3 polyunsaturated fatty acid highly allowed to breed (sex ratio 1:3). On the following morn-
enriched in the brain, is vital for proper brain develop- ing the vaginal smears were taken to confirm mating.
ment and function [26]. Micronutrient insufficiency or Vaginal smears were taken on a clean microscope slide
imbalance during pregnancy can compromise brain using a cotton bud dipped in saline. The slides were
development and function. Our studies in young schizo- examined under a microscope at 10 magnification.
phrenic patients have also shown altered micronutrients The sperm positive smear was considered a result of suc-
and DHA at the onset of psychosis implicating a neuro- cessful mating and considered day 0 of gestation. The
developmental pathophysiology in schizophrenia [27]. pregnant dams were housed individually (in polypropyl-
This has led us recently to describe that in one carbon ene cages of 29  22  14 cm dimensions containing rice
metabolic pathway, membrane phospholipids are major husk as bedding material) and allowed to deliver nor-
methyl group acceptors and reduced DHA levels may mally. Animals receiving Vitamin B12 deficient diets
result in diversion of methyl groups towards DNA ulti- were kept in special cages to prevent coprophagy.
mately resulting in DNA methylation [28]. Reports sug- Out of 60 females, 47 females became pregnant and
gest that many neurological disorders like childhood divided randomly into six dietary groups. All dams were
cognitive dysfunction, schizophrenia which become delivered by C section on d20 of gestation. Two pups
apparent after birth have their origin during fetal life from each group were randomly selected and analyzed
[29–32]. for organ weights and biochemical parameters.
We hypothesize that altered maternal intake and
metabolism of folic acid, vitamin B12 and omega 3 fatty 2.3. Diets
acids in one carbon metabolism and induction of oxida-
tive stress during pregnancy may lead to reduced levels The composition of the control and the treatment
of brain LCPUFA in the offspring at birth which may diets was as per AIN 93 purified diets for laboratory
have implications for neurodevelopmental disorders in rodents [33]. Protein level in the control and treatment
later life. The present study for the first time examines diets was 18%. Total of six isocaloric treatment diets
the effect of (1) imbalance of maternal micronutrients were formulated. Four diets were formulated for exam-
(folic acid and vitamin B12) and (2) maternal omega 3 ining the effects of two (i.e. 2 and 8 mg folic acid/kg diet)
fatty acid supplementation in the above diets on brain different levels of folic acid during pregnancy both in the
fatty acids and oxidative stress parameters in the off- presence and absence of vitamin B12. In addition, two
spring at birth. more diets were formulated to examine the effects of
omega 3 fatty acid (DHA + EPA) supplementation on
both the vitamin B12 deficient groups. Vitamin B12 defi-
2. Materials and methods ciency was obtained exclusively through dietary means.
The diets contained the following ingredients (g/kg
All experimental procedures were in accordance with diet) (casein, 200; corn starch, 398; dextrinized starch,
guidelines of Institutional Animal Ethics Committee 132; sucrose, 100; fiber, 50; mineral mixture, 35; vitamin
(IAEC). The institute is recognized to undertake exper- mixture, 10; cystine, 3; choline bitartarate, 2.5; tertiary
iments on animals as per the committee for the purpose butyl hydroxyl quinone, 0.014; soyabean oil, 70).
66 S. Roy et al. / Brain & Development 34 (2012) 64–71

Vitamin free casein was used for all treatment diets. The with a least count of 0.001 g and maximum capacity of
mineral mixture composed of the following ingredients 220 g. These brains were immediately snap frozen in
(g/kg mixture): calcium carbonate, 357; potassium phos- liquid nitrogen and stored at 80 °C for biochemical
phate, 196; potassium citrate, 70.78; sodium chloride, estimations. The relative brain weights were expressed
78; potassium sulphate, 46.6; magnesium oxide, 24; fer- in relation to the pup weights i.e. (absolute brain
ric citrate, 6.06; zinc carbonate, 1.65; manganous car- weight/pup weight  100).
bonate, 0.63; cupric carbonate, 0.3; potassium iodate,
0.01; sodium selenate, 0.01; ammonium paramolybdate, 2.6. Analysis of fatty acids
0.007; sodium metasilicate, 1.45; chromium potassium
sulphate, 0.275; lithium chloride, 0.01; boric acid, 0.08; Whole brains of the pups were homogenized in cold
sodium fluoride, 0.06; nickel carbonate, 0.03; ammo- phosphate buffer saline, pH 7.5 using a Teflon glass
nium vanadate, 0.006 and sucrose, 221.02. The vitamin homogenizer on ice. The homogenate was centrifuged at
mixture composed of the following ingredients (g/kg 10,000 rpm at 4 °C for 20 min. The membranes were
mixture): nicotinic acid, 3; calcium pantothenate, 1.6; diluted in 2 ml phosphate buffer saline and stored at
pyridoxine–HCl, 0.7; thiamin–HCl, 0.6; riboflavin, 0.6; 20 °C. Fatty acids were analyzed as methyl esters by
D-Biotin, 0.02; vitamin B12 (in 0.1% Mannitol), 2.5; vita- the method as recently described by us [37]. Briefly,
min E, 15; vitamin A, 0.8; vitamin D-3, 0.25; vitamin K, transesterification of the phospholipid fraction was
0.075; folic acid, 0.2 (control) and sucrose 974.655, was carried out using HCl–methanol. Methyl esters were
used to make total weight of the vitamin mixture to separated and quantified using a Perkin-Elmer gas chro-
1 kg. matograph (SD 2330, 30 m capillary column, Supelco,
Thus there were a total of six groups, control PA, USA). Helium was used as carrier gas at 1 ml/min.
(NLFolate + NLB12 i.e. normal folate, normal B12); Oven temperature was held at 150 °C for 10 min pro-
NLFolate + noB12 (normal folate, B12 deficient); NLFo- grammed to rise from 150 to 220 °C at 10 °C/min and at
late + noB12 + omega 3 supp (normal folate, B12 defi- 220 °C for 10 min. The detector temperature was 275 °C
cient, omega 3 supplemented); HFolate + NLB12 and the injector temperature was 240 °C. Retention times
(High folate, normal B12); HFolate + noB12 (High and peak areas were automatically computed. Individual
folate, B12 deficient) and HFolate + noB12 + omega 3 fatty acids were identified by comparison of peaks with
supp (High folate, B12 deficient, omega 3 supplemented). peaks of standard fatty acid methyl esters (Sigma,
The lowest level i.e. 2 mg/kg represents the normal USA). The data included is on only critically important
level of folic acid used in the control diet as per the cur- long chain polyunsaturated fatty acids and major fatty
rent AIN 93 guidelines while 8 mg/kg is roughly four acids. The omega 3 fatty acids included alpha linolenic
times the requirement of a normal rat. This is in accor- acid, eicosapentaenoic acid and docosahexaenoic acid
dance with the fact that folic acid requirement for while omega-6 fatty acids included linolenic acid, gamma
Indian pregnant woman is set at 400 lg/d which is four linolenic acid, dihomogammalinolenic acid, docosapen-
times the requirement of a non-pregnant woman. The taenoic acid and arachidonic acid. The monounsaturated
level of omega 3 fatty acid supplementation was chosen fatty acids included myristoleic acid, palmitoleic acid,
to have an omega 6/omega 3 ratio of 1:1 which is consid- oleic acid and nervonic acid. Saturated fatty acids
ered to be the ideal ratio [34]. included myristic acid, palmitic acid and stearic acid.
All diets contained tertiary butyl hydroquinone
(TBHQ) to prevent oxidation [33,35,36]. Diets were 2.7. Lipid peroxidation measurements (dam plasma and
stored at refrigeration temperatures and fresh diet was pup brain MDA levels)
provided daily.
Malondialdehyde (MDA) levels were estimated from
2.4. Observations recorded dam plasma and pups brain using kit from Bioxytech
MDA-586 kit (Oxis International, USA). Briefly, thio-
During pregnancy, dam weights were recorded at 0, barbituric acid reacts with malondialdehyde to form
7, 14 and 20 d to obtain weight gains. On d20 of gesta- pink color, and the absorbance was measured at
tion the litter weight and size was recorded in each 586 nm. Tetramethoxypropane is used as a standard.
group. Brain MDA concentration is expressed as nmol/ml.

2.5. Brain weights 2.8. Statistical methods

The pups were cut open from the gestational sac and Litter means were used as the unit of analysis. Values
weighed before dissecting. Then the pups were dissected are mean ± SD. The data were analyzed using SPSS/
to collect the whole brain and the absolute weight of the PC + package (Version 11.0, Chicago, IL). Mean values
brain was recorded on a Schimadzu electronic balance of the estimates of various parameters for the treatment
S. Roy et al. / Brain & Development 34 (2012) 64–71 67

groups were compared with those of control group at MDA from the HFolate + noB12 group was also higher
conventional levels of significance, using least signifi- (p < 0.01) as compared to control and HFolate + NLB12
cance difference estimated from one way analysis of var- groups (Fig. 1).
iance (ANOVA).
3.5. Dam plasma fatty acids
3. Results
DHA and AA were comparable in the HLFolate +
3.1. Feed intake NLB12 group as compared to control. However, DHA
levels in the HLFolate + noB12 group was lower
The daily feed intake was (19.44 ± 1.86 g) for control; (p < 0.05) as compared to control (Fig. 1). Supplementa-
(17.80 ± 3.12 g) for NLFolate + noB12; (18.22 ± 2.14 g) tion of omega 3 fatty acids to the NLFolate + noB12 and
for HFolate + NLB12 and (18.49 ± 3.72 g) for HFo- HLFolate + noB12 groups improved levels of DHA and
late + noB12 groups suggesting that imbalance in maternal reduced levels of AA (p < 0.01 for both) as compared to
micronutrients (folic acid and vitamin B12) did not affect control. Further, DHA levels in HLFolate + noB12 +
feed intake. In contrast, feed intake in both the omega 3 omega 3 supp was higher and AA levels were lower as
fatty acid supplemented groups was lower (p < 0.01) i.e. compared to HLFolate + noB12 group.
NLFolate + noB12 + omega 3 supp (14.91 ± 1.33 g) and
HFolate + noB12 + omega 3 supp (15.32 ± 2.15 g) than 3.6. Pup brain fatty acids
control. Further feed intake in omega 3 fatty acid supple-
mented groups were also lower than their respective B12 In the pup brain DHA levels were lower in both the
deficient groups (NLFolate + noB12 vs. NLFolate +noB NLFolate + noB12 and HLFolate + noB12 groups (p <
12 + omega 3 supp, p < 0.05) and (HFolate + noB12 vs. 0.01 for both) as compared to control (Fig. 1).
HFolate + noB12 + omega 3 supp, p < 0.05). Reductions in DHA (p < 0.05) were also seen in the
HLFolate + NLB12 group as compared to control. In
3.2. Reproductive performance contrast, both NLFolate + noB12 + omega supp and
HLFolate + noB12 + omega supp groups showed
Weight gain of dams during pregnancy was similar in improved DHA levels (p < 0.01 for both). However, ara-
all the groups i.e. control (106.5 ± 19.97), NLFolate + chidonic acid levels were reduced (p < 0.05) as compared
noB12 (111.75 ± 20.10), HFolate + NLB12 (103.00 ± to control and their respective B12 deficient groups.
37.78) and HFolate + noB12 (113.50 ± 26.30). Further
supplementation of omega 3 fatty acids to the vitamin 4. Discussion
B12 deficient groups was also similar i.e. NLFolate +
noB12 + omega 3 supp (107.87 ± 13.88) and HFolate + This is the first report which has examined the effect of
noB12 + omega 3 supp (103.75 ± 19.36). The pup weight imbalance of maternal micronutrients (folic acid and
was comparable between all treatment groups i.e. vitamin B12) and omega 3 fatty acid supplementation
control (3.51 ± 0.92), NLFolate + noB12 (3.74 ± 0.61), on maternal oxidative stress marker (malondialdehyde)
HFolate + NLB12 (3.61 ± 1.23), HFolate + noB12 and pup brain LCPUFA levels on d20 of gestation.
(3.36 ± 0.51), NLFolate + noB12 + omega 3 supp (3.15 ± Our results indicate (1) imbalance in maternal micronu-
0.81) and HFolate + noB12 + omega 3 supp (3.40 ±0.41). trients like folic acid and vitamin B12 or omega 3 fatty
acid supplementation did not affect weight gain during
3.3. Relative brain weights pregnancy. (2) Maternal folic acid supplementation in
the presence of vitamin B12 did not alter oxidative stress
There was no difference in relative brain weights levels in the dam and pup (3) however, maternal folic acid
of pups among all the groups i.e. control (0.04 ± supplementation in the absence of vitamin B12 increased
0.007), NLFolate + noB12 (0.04 ± 0.005), HFolate + oxidative stress levels both in the dam and pup (4) mater-
NLB12 (0.04 ± 0.008), HFolate + noB12 (0.04 ± 0.008), nal folic acid supplementation both in the presence and
NLFolate + noB12 + omega 3 supp (0.04 ± 0.01) and absence of vitamin B12 reduced brain DHA levels in the
HFolate + noB12 + omega 3 supp (0.04 ± 0.001). pup (5) Maternal omega 3 fatty acids supplementation
to the vitamin B12 deficient groups resulted in signifi-
3.4. Dam plasma and pup brain MDA levels cantly higher brain DHA and lower AA levels.
In our study there was no difference in the total
Dams plasma MDA levels from the HFolate + noB12 weight gain of animals among all the groups. Our results
group was higher (p < 0.01) as compared to control, are similar to those by Achon et al. [38] who showed that
NLFolate + noB12 and HFolate + NLB12 groups folic acid supplementation at 40 mg/kg diet did not
(Fig. 1). However, plasma MDA levels in dams from the affect the gestation outcome. There are no reports which
other groups was comparable to control. Pup brain have examined effects of folic acid supplementation in
68 S. Roy et al. / Brain & Development 34 (2012) 64–71

Fig. 1. Maternal plasma and pup brain malondialdehyde and fatty acid levels. *p < 0.05; **p < 0.01; compared to control; #p < 0.05; ##p < 0.01;
compared to NLFolate, noB12 $p < 0.05; $$p < 0.01; compared to HFolate, normal B12; @p < 0.05; @@p < 0.01; compared to HFolate,
9 99
noB12; xp < 0.05; xxp < 0.01 compared to NLFolate, noB12, omega 3 supp.; control – normal folate, normal B12; NLFolate, noB12 – normal
folate, B12 deficient; NLFolate, noB12, omega 3 supp – normal folate, B12 deficient, omega 3 supplemented HFolate, normal B12 – high folate, normal
B12; HFolate, noB12 – high folate, B12 deficient; HFolate, noB12, omega 3 supp – high folate, B12 deficient, omega 3 supplemented.

absence of vitamin B12 on birth outcome. However, in a was no difference in the body weights before and after
recent study when male rats were supplemented with supplementation [39]. In our study, omega 3 fatty acid
folic acid in the absence of vitamin B12 deficiency there did not influence the weight gain. Others also report that
S. Roy et al. / Brain & Development 34 (2012) 64–71 69

varying ratios of omega 3 fatty:omega 6 fatty acids dur- reactions would further promote decomposition of
ing pregnancy on a control diet do not show any effect polyunsaturated fatty acids [25].
on weight gain [40]. The present study for the first time reports lower pup
There was no difference in litter weight or size in all brain DHA levels in both the maternal vitamin B12 defi-
groups due to altered levels of maternal micronutrients cient groups irrespective of the level of folic acid. We have
in this study. Our earlier studies examining the effects earlier reported that maternal folic acid supplementation
of folic acid supplementation (8 mg/kg) at marginal pro- at marginal protein levels reduced brain DHA levels in
tein diet during pregnancy also found no change in litter the adult offspring [20,54]. Altered intake/metabolism
weight or litter size [41]. In contrast a study examining of folic acid and vitamin B12 may induce oxidative stress
the effect of supplementation of excess folic acid leading to increased MDA from LCPUFA.
(40 mg/kg) during pregnancy showed reductions in body Vitamin B12 and folate are essential for methylation of
weight of the fetuses as compared to control [38]. In our homocysteine to methionine and for synthesis of s-aden-
study when omega 3 fatty acid was given along with osyl methionine. s-Adenosyl methionine is involved in
excess folic acid in the absence of vitamin B12 there the metabolism of proteins, phospholipids and neuro-
was no change in pup weight. Maternal diets containing transmitters [28]. This also can reduce levels of glutathi-
varying ratios of omega 3 fatty:omega 6 fatty acids [40], one, a major antioxidant during pregnancy in mother
or docosahexaenoic acid and eicosapentaenoic acid [42] and fetus. In the liver, phosphatidylcholine (PC) is also
have also shown no alterations in pup weights at birth. synthesized by the sequential methylation of phosphati-
However, when pregnant women were given 200 mg dylethanolamine (PE), which is catalyzed by phosphati-
DHA supplements from gestation week 21 until the dylethanolamine N-methyltransferase (PEMT) [55]. In
end of third month of lactation, their infants had lower vitamin B deficiency, the amounts of methionine
body weight [43]. In contrast maternal consumption of available for phosphatidylcholine synthesis is reduced
omega 3 fatty acids during pregnancy have reported and a lower methylation of phosphatidylethanolamine
an increase in birth weight [44–47]. to phosphatidylcholine has been reported [56]. A defect
It is known that the brain is vulnerable to free radical in methylation process is hypothesized to be the biochem-
attack and lipid peroxidation (LPO) [48]. There are no ical basis of neuropsychiatric manifestations of vitamin
studies which have examined the effect of imbalance of B12 deficiency [57].
maternal micronutrients or omega 3 fatty acid supple- Folic acid and vitamin B12 are the major determi-
mentation to these diets on brain plasma MDA levels, nants of one carbon metabolism which alter levels of
an index of lipid peroxidation. In our study maternal vita- both homocysteine and oxidative stress (MDA). One
min B12 deficiency did not increase plasma MDA levels in of the major functions of one carbon metabolism is also
the dams. However, excess folic acid in the absence of to regulate the membrane phospholipids levels especially
vitamin B12 increased plasma MDA levels. A recent study DHA through the transfer of methyl groups. Therefore
in adult rats reports increased malondialdehyde (MDA) altered intake/metabolism of maternal micronutrients
levels in kidney tissue of vitamin B6 deficient male rats may alter levels of DHA (Fig. 2).
[49] while folic acid and folic acid + vitamin B12 supple- Supplementing the vitamin B12 deficient groups with
mentation in adult rats has also been shown to reduce sys- omega 3 fatty acids in our study resulted in higher levels
temic oxidative stress in male rats [50]. Patients with of brain DHA levels but reduced AA levels. A recent study
severe neonatal cobalamin (vitamin B12) deficiency have
been reported to have elevated intracellular ROS content
[51]. It has been suggested that dietary vitamin B12 folic
acid, and omega 3 PUFAs may influence plasma and
neuronal levels of oxidative stress, that may ultimately
determine the susceptibility of an individual to develop
neurodegenerative disorders [52].
It is known that LCPUFA is susceptible for
degradation due to increased oxidative stress [53].
The imbalances in micronutrients often lead to
increased oxidative stress through the generation of
oxygen free radicals which interact with polyunsatu-
rated fatty acids in membranes or lipoproteins.
Reactive oxygen species attack the double bonds of
polyunsaturated fatty acids and initiate chain reactions
leading to lipid peroxide formation. Our earlier studies
in women with pregnancy complications like pre- Fig. 2. Altered maternal micronutrient interactions leading to impaired
eclampsia also suggest that increased peroxidative brain development in the offspring.
70 S. Roy et al. / Brain & Development 34 (2012) 64–71

has shown that early supplementation of maternal diet [3] Owens S, Fall CH. Consequences of poor maternal micronutri-
with omega 3 fatty acids supplied as EPA and DHA is tion before and during early pregnancy. Trans R Soc Trop Med
Hyg 2008;102:103–4.
efficient in increasing omega 3 in brain of the neonate [4] Pitkin RM. Folate and neural tube defects. Am J Clin Nutr
[42]. Several neurodevelopmental disorders like schizo- 2007;85:285S–8S.
phrenia [58] and ADHD [59] are associated with increased [5] Indian Council of Medical Research. Evaluation of the national
oxidative stress. However there is no information when nutritional anaemia prophylaxis programme, an ICMR task force
and how oxidative stress triggered by an etiopathogenetic study. Central Electric, New Delhi; 1989.
[6] Food and Drug Administration. Food standards: amendment of
factor(s) affects the early neurodevelopment and contrib- standards of identity for enriched grain products to require
utes to onset of psychopathology and its course and treat- addition of folic acid. Final rule. 21 CFR Parts 136, 137, and 139.
ment throughout life. Fed Regist 1996;61:8781–7.
Our results suggest that oxidative stress is most likely [7] Freire WB, Hertrampf E, Cortes F. Effect of folic acid fortifica-
triggered due to imbalance in maternal micronutrients tion in Chile: preliminary results. Eur J Pediatr Surg
2000;10(Suppl.):42–3.
like folic acid and vitamin B12 during fetal growth. This [8] Health Canada. Regulations amending the Food and Drug
may continue through adolescence thereby contributing Regulations (1066). Can Gaz Part 1 1997;131:3702–37.
to neurodevelopmental deficits and onset of illness. [9] Metz J, Sikaris KA, Maxwell EL, Levin MD. Changes in serum
These micronutrients are involved in 1-carbon metabo- folate concentrations following voluntary food fortification in
lism and act as co-factors or molecular donors for Australia. Med J Aust 2002;176:90–1.
[10] Bower C, Ryan A, Rudy E, Miller M. Trends in neural tube
epigenetic processes such as DNA methylation, which defects in Western Australia. Aust NZ J Public Health
modulate gene expression, and therefore cellular prolif- 2002;26:150–1.
eration and apoptosis, in early pregnancy [3]. There is a [11] De Wals P, Rusen ID, Lee NS, Morin P, Niyonsenga T. Trend in
need to determine the underlying mechanisms of action prevalence of neural tube defects in Quebec. Birth Defects Res A
of these nutrients, including effects mediated via epige- Clin Mol Teratol 2003;67:919–23.
[12] Gucciardi E, Pietrusiak MA, Reynolds DL, Rouleau J. Incidence
netic modifications. Our lab has therefore undertaken of neural tube defects in Ontario, 1986–1999. CMAJ
studies to examine the epigenetic changes occurring at 2002;167:237–40.
the gene specific level for antioxidant enzymes (SOD [13] Persad VL, Van den Hof MC, Dubé JM, Zimmer P. Incidence of
and GPx) and neurotrophins like brain derived neuro- open neural tube defects in Nova Scotia after folic acid fortifi-
trophic factor which is known to be regulated by levels cation. CMAJ 2002;167:241–5.
[14] Shane B. Folate fortification: enough already? Am J Clin Nutr
of DHA [60,61] and these findings may have important 2003;77:8–9.
implications in neurodevelopmental disorders. [15] Williams LJ, Mai CT, Edmonds LD, Shaw GM, Kirby RS,
This study for the first time indicates that imbalance Hobbs CA, et al. Prevalence of spina bifida and anencephaly
in maternal micronutrients reduces the brain DHA lev- during the transition to mandatory folic acid fortification in the
els in the offspring at birth probably through the United States. Teratology 2002;66:33–9.
[16] Kim YI. Will mandatory folic acid fortification prevent or
observed increase in oxidative stress. Supplementation promote cancer? Am J Clin Nutr 2004;80:1123–8.
with omega 3 fatty acids to these imbalanced diets [17] Carmel R. Efficacy and safety of fortification and supplementa-
improved the brain DHA levels. Our data has implica- tion with vitamin B12: biochemical and physiological effects. Food
tions for several neurodevelopmental disorders since Nutr Bull 2008;29:S177–87.
micronutrients and DHA are important modulators [18] Smith SE, Kinney HC, Swoboda KJ, Levy HL. Subacute
combined degeneration of the spinal cord in cblC disor-
for neural functioning. Therefore it would be of inter- der despite treatment with B12. Mol Genet Metab 2006;88:
est to examine whether the observed alterations in 138–45.
maternal micronutrients and the consequential reduc- [19] Yajnik CS, Deshpande SS, Lubree HG, Naik SS, Bhat DS,
tions in brain fatty acids in the offspring’s are due to Uradey BS, et al. Vitamin B12 deficiency and hyperhomocystein-
the altered methylation and expression of PEMT. emia in rural and urban Indians. J Assoc Physicians India
2006;54:775–82.
Future studies need to also address the associated [20] Rao S, Joshi S, Kale A, Hegde M, Mahadik S. Maternal folic acid
changes in brain structure and function during the supplementation to dams on marginal protein level alters brain
adult life. fatty acid levels of their adult offspring. Metab Clin Exp
2006;55:628–34.
Conflict of interest statement [21] Dangat K, Mehendale S, Yadav H, Kilari AS, Kulkarni AV,
Taralekar VS, et al. Long chain polyunsaturated fatty acid
composition of breast milk in pre-eclamptic mothers. Neonatol-
None. ogy 2009;97:190–4.
[22] Joshi SR, Mehendale SS, Dangat KD, Kilari AS, Yadav HR,
References Taralekar VS. High maternal plasma antioxidant concentrations
associated with preterm delivery. Ann Nutr Metab 2008;53:
[1] Barker DJ. Developmental origins of adult health and disease. J 276–82.
Epidemiol Community Health 2004;58:114–5. [23] Kilari AS, Mehendale SS, Dangat KD, Yadav HR, Kulakarni
[2] Christian P, Stewart CP. Maternal micronutrient deficiency, fetal AV, Dhobale MV, et al. Long chain polyunsaturated fatty
development, and the risk of chronic disease. J Nutr 2010;140: acids in mothers and term babies. J Perinat Med 2009;37:
437–45. 513–8.
S. Roy et al. / Brain & Development 34 (2012) 64–71 71

[24] Kilari AS, Mehendale SS, Dangat KD, Yadav HR, Gupta A, nancy and lactation lower BMI in late infancy? J Perinat Med
Taralekar VS, et al. Long chain polyunsaturated fatty acids in 2007;35:295–300.
mothers of preterm babies. J Perinat Med 2009;37:513–8. [44] Olsen SF, Hansen HS, Sørensen T, et al. Hypothesis: dietary (N-
[25] Mehendale S, Kilari A, Dangat K, Taralekar V, Mahadik S, Joshi 3)-fatty acids prolong gestation in human beings. Prog Clin Biol
S. Fatty acids, antioxidants, and oxidative stress in pre-eclampsia. Res 1987;242:51–6.
Int J Gynaecol Obstet 2008;100:234–8. [45] Olsen SF, Secher NJ. A possible preventive effect of low-dose fish
[26] Innis SM. Dietary omega 3 fatty acids and the developing brain. oil on early delivery and pre-eclampsia: indications from a 50-
Brain Res 2008;1237:35–43. year-old controlled trial. Br J Nutr 1990;64:599–609.
[27] Kale A, Joshi S, Naphade N, et al. Opposite changes in [46] Sattar N, Berry C, Greer IA. Essential fatty acids in relation to
predominantly docosahexaenoic acid (DHA) in cerebrospinal pregnancy complications and fetal development. Br J Obstet
fluid and red blood cells from never-medicated first-episode Gynaecol 1998;105:1248–55.
psychotic patients. Schizophr Res 2008;98:295–301. [47] Smuts CM, Huang M, Mundy D, et al. A randomized trial of
[28] Kale A, Naphade N, Sapkale S, Kamaraju M, Pillai A, Joshi S, docosahexaenoic acid supplementation during the third trimester
et al. Reduced folic acid, vitamin B12 and docosahexaenoic acid of pregnancy. Obstet Gynecol 2003;101:469–79.
and increased homocysteine and cortisol in never-medicated [48] Ansari MA, Roberts KN, Scheff SW. Oxidative stress and
schizophrenia patients: Implications for altered one-carbon modification of synaptic proteins in hippocampus after traumatic
metabolism. Psychiatry Res 2010;175:47–53. brain injury. Free Radic Biol Med 2008;45:443–52.
[29] Cannon TD, Yolken R, Buka S, et al. Collaborative Study Group [49] Keles M, Al B, Gumustekin K, et al. Antioxidative status and
on the Perinatal Origins of Severe Psychiatric Disorders. lipid peroxidation in kidney tissue of rats fed with vitamin B(6)-
Decreased neurotrophic response to birth hypoxia in the etiology deficient diet. Ren Fail 2010;32:618–22.
of schizophrenia. Biol Psychiatry 2008;64:797–802. [50] Majumdar S, Mukherjee S, Maiti A, et al. Folic acid or
[30] Eriksson JG. The fetal origins hypothesis-10 years on. BMJ combination of folic acid and vitamin B(12) prevents short-term
2005;330:1096–7. arsenic trioxide-induced systemic and mitochondrial dysfunction
[31] Rees S, Harding R. Brain development during fetal life: influences and DNA damage. Environ Toxicol 2009;24:377–87.
of the intra-uterine environment. Neurosci Lett 2004;361:111–4. [51] Richard E, Alvarez-Barrientos A, Pérez B, et al. Methylmalonic
[32] Rehn AE, Van Den Buuse M, Copolov D, et al. An animal model acidaemia leads to increased production of reactive oxygen species
of chronic placental insufficiency: relevance to neurodevelopmen- and induction of apoptosis through the mitochondrial/caspase
tal disorders including schizophrenia. Neuroscience pathway. J Pathol 2007;213:453–61.
2004;129:381–91. [52] Das UN. Folic acid and polyunsaturated fatty acids improve
[33] Reeves P, Nielsen F, Fahey Jr G. AIN-93 purified diets for cognitive function and prevent depression, dementia, and Alzhei-
laboratory rodents: final report of the American Institute of mer’s disease – but how and why? Prostaglandins Leukot Essent
Nutrition Ad Hoc Writing Committee on the reformulation of the Fatty Acids 2008;78:11–9.
AIN-76A rodent diet. J Nutr 1993;123:1939–51. [53] Song JH, Fujimoto K, Miyazawa T. Polyunsaturated (n-3) fatty
[34] Simopoulos AP. Essential fatty acids in health and chronic acids susceptible to peroxidation are increased in plasma and
disease. Am J Clin Nutr 1999;70:560S–9S. tissue lipids of rats fed docosahexaenoic acid-containing oils. J
[35] Fritsche KL, Johnston PV. Rapid autoxidation of fish oil in diets Nutr 2000;130:3028–33.
without added antioxidants. J Nutr 1988;118:425–6. [54] Joshi S, Rao S, Girigosavi S, et al. Differential effects of fish oil
[36] Gonzalez MJ, Gray JI, Schemmel RA, et al. Lipid peroxidation and folate supplementation during pregnancy in rats on cognitive
products are elevated in fish oil diets even in the presence of added performance and serum glucose level in their offspring. Nutrition
antioxidants. J Nutr 1992;122:2190–5. 2004;20:465–72.
[37] Ranjekar PK, Hinge A, Hegde MV, et al. Decreased antioxidant [55] Vance JE. Phosphatidylserine and phosphatidylethanolamine in
enzymes and membrane essential polyunsaturated fatty acids in mammalian cells: two metabolically related aminophospholipids.
schizophrenic and bipolar mood disorder patients. Psychiatry Res J Lipid Res 2008;49:1377–87.
2003;121:109–22. [56] Akesson B, Fehling C, Jägerstad M. Lipid composition and
[38] Achon M, Alonso-Aperte E, Reyes L, et al. High dose folic acid metabolism in liver and brain of vitamin B12-deficient rat
supplementation in rats: effects on gestation and methionine cycle. sucklings. Br J Nutr 1979;41:263–74.
Br J Nutr 2000;83:177–83. [57] Bottiglieri T. Folate, vitamin B12, and neuropsychiatric disorders.
[39] Min H. Effects of dietary supplementation of high-dose folic acid Nutr Rev 1996;54:382–90.
on biomarkers of methylating reaction in vitamin B(12)-deficient [58] Arvindakshan M, Sitasawad S, Debsikdar V, et al. Essential
rats. Nutr Res Pract 2009;3:122–7. polyunsaturated fatty acid and lipid peroxide levels in never-
[40] Jen KL, Church MW, Wang C, et al. Perinatal n-3 fatty acid medicated and medicated schizophrenia patients. Biol Psychiatry
imbalance affects fatty acid composition in rat offspring. Physiol 2003;53:56–64.
Behav 2009;98:17–24. [59] Richardson AJ. Omega 3 fatty acids in ADHD and related
[41] Joshi S, Rao S, Golwilkar A, et al. Fish oil supplementation of neurodevelopmental disorders. Int Rev Psychiatry
rats during pregnancy reduces adult disease risks in their 2006;18:155–72.
offspring. J Nutr 2003;133:3170–4. [60] Wu A, Ying Z, Gomez-Pinilla F. Dietary omega 3 fatty acids
[42] Destaillats F, Joffre C, Acar N, Joffre F, et al. Differential effect of normalize BDNF levels, reduce oxidative damage, and counteract
maternal diet supplementation with alpha-linolenic acid or n-3 learning disability after traumatic brain injury in rats. J Neuro-
long-chain polyunsaturated fatty acids on glial cell phosphatidyl- trauma 2004;21:1457–67.
ethanolamine, phosphatidylserine fatty acid profile in neonate rat [61] Wu A, Ying Z, Gomez-Pinilla F. Docosahexaenoic acid dietary
brains. Nutr Metab (Lond) 2010;7:2. supplementation enhances the effects of exercise on synaptic
[43] Lucia Bergmann R, Bergmann K, Haschke-Becher E, et al. Does plasticity and cognition. Neuroscience 2008;155:751–9.
maternal docosahexaenoic acid supplementation during preg-

View publication stats

You might also like